+ All Categories
Home > Documents > Chapter 7 0 Exoribonucleases - Liang Tong

Chapter 7 0 Exoribonucleases - Liang Tong

Date post: 23-Mar-2022
Category:
Upload: others
View: 3 times
Download: 0 times
Share this document with a friend
26
Chapter 7 5 0 -3 0 Exoribonucleases Jeong Ho Chang, Song Xiang, and Liang Tong Contents 7.1 Introduction .............................................................................. 168 7.2 Sequence Conservation of the XRNs ................................................... 168 7.3 5 0 -3 0 Exonuclease Activity of XRNs .................................................... 170 7.4 Functions of Xrn1 ........................................................................ 172 7.4.1 Functions of Xrn1 in RNA Degradation and Turnover ........................ 172 7.4.2 Functions of Xrn1 in RNA Maturation ......................................... 174 7.4.3 Functions of Xrn1 in DNA Recombination and Chromosome Stability ...... 174 7.4.4 Other Functions of Xrn1 ......................................................... 175 7.5 Functions of Xrn2/Rat1 .................................................................. 175 7.5.1 Functions of Xrn2/Rat1 in RNA Processing and Degradation ................. 176 7.5.2 Functions of Xrn2/Rat1 in RNA Polymerase Transcription Termination ..... 176 7.5.3 Other Functions of Xrn2/Rat1 ................................................... 177 7.6 Protein Partners of XRNs ................................................................ 178 7.7 Functions of XRNs in Plants and Other Organisms .................................... 179 7.8 Overall Structure of Xrn2/Rat1 .......................................................... 179 7.9 Active Site of Xrn2/Rat1 ................................................................ 181 7.10 Structure of the Rat1-Rai1 Complex .................................................... 182 7.11 Rai1/Dom3Z and RNA 5 0 -End Capping ................................................ 183 7.12 The 5 0 -3 0 Exoribonuclease Rrp17 ....................................................... 185 7.13 RNase J1/CPSF-73 ....................................................................... 185 7.14 Perspectives .............................................................................. 186 References ....................................................................................... 186 J.H. Chang • L. Tong (*) Department of Biological Sciences, Columbia University, New York, NY 10027, USA e-mail: [email protected] S. Xiang Department of Biological Sciences, Columbia University, New York, NY 10027, USA Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China A.W. Nicholson (ed.), Ribonucleases, Nucleic Acids and Molecular Biology 26, DOI 10.1007/978-3-642-21078-5_7, # Springer-Verlag Berlin Heidelberg 2011 167
Transcript

Chapter 7

50-30 Exoribonucleases

Jeong Ho Chang, Song Xiang, and Liang Tong

Contents

7.1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168

7.2 Sequence Conservation of the XRNs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168

7.3 50-30 Exonuclease Activity of XRNs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170

7.4 Functions of Xrn1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172

7.4.1 Functions of Xrn1 in RNA Degradation and Turnover . . . . . . . . . . . . . . . . . . . . . . . . 172

7.4.2 Functions of Xrn1 in RNA Maturation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174

7.4.3 Functions of Xrn1 in DNA Recombination and Chromosome Stability . . . . . . 174

7.4.4 Other Functions of Xrn1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175

7.5 Functions of Xrn2/Rat1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175

7.5.1 Functions of Xrn2/Rat1 in RNA Processing and Degradation . . . . . . . . . . . . . . . . . 176

7.5.2 Functions of Xrn2/Rat1 in RNA Polymerase Transcription Termination . . . . . 176

7.5.3 Other Functions of Xrn2/Rat1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177

7.6 Protein Partners of XRNs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178

7.7 Functions of XRNs in Plants and Other Organisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179

7.8 Overall Structure of Xrn2/Rat1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179

7.9 Active Site of Xrn2/Rat1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 181

7.10 Structure of the Rat1-Rai1 Complex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182

7.11 Rai1/Dom3Z and RNA 50-End Capping . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 183

7.12 The 50-30 Exoribonuclease Rrp17 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185

7.13 RNase J1/CPSF-73 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185

7.14 Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 186

J.H. Chang • L. Tong (*)

Department of Biological Sciences, Columbia University, New York, NY 10027, USA

e-mail: [email protected]

S. Xiang

Department of Biological Sciences, Columbia University, New York, NY 10027, USA

Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai

Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China

A.W. Nicholson (ed.), Ribonucleases, Nucleic Acids and Molecular Biology 26,

DOI 10.1007/978-3-642-21078-5_7, # Springer-Verlag Berlin Heidelberg 2011

167

Abstract The 50-30 exoribonucleases have important functions in RNA processing,

RNA degradation, RNA interference, transcription, and other cellular processes.

The Xrn1 and Xrn2/Rat1 family of enzymes are the best characterized 50-30

exoribonucleases, and there has been significant recent progress in the understand-

ing of their structure and function. Especially, the first structural information on

Rat1 just became available. Other 50-30 exoribonucleases have been identified

recently, including yeast Rrp17 and B. subtilis RNase J1, the first enzyme with

50-30 exoribonuclease activity found in prokaryotes. This review will summarize

our current understanding of these enzymes, focusing on their sequence conserva-

tion, molecular structure, biochemical and cellular functions.

7.1 Introduction

Exoribonucleases are involved in RNA processing, RNA degradation, RNA inter-

ference, transcription, modulation of gene expression, antiviral defense, and other

cellular processes. These enzymes can be simply classified based on the direction of

their activity, hence 50-30 or 30-50 exoribonucleases. While a large number of 30-50

exoribonucleases have been identified, in bacteria and eukaryotes (Zuo and

Deutscher 2001) (see also Chap. 8), few 50-30 exoribonucleases are currently

known. The best characterized 50-30 exoribonucleases are the Xrn1/Xrn2 family

of enzymes (to be referred to as XRNs here), which have only been found in

eukaryotes.

Recently, Rrp17 was identified as another 50-30 exoribonuclease, with an impor-

tant role in the 50-end processing of pre-ribosomal RNAs (Oeffinger et al. 2009).

Several enzymes that possess both endo- and 50-30 exoribonuclease activity have

also been reported, including B. subtilis RNase J1 (Condon 2010), the first enzyme

with 50-30 exoribonuclease activity found in prokaryotes (see also Chap. 10). RNaseJ1 is structurally homologous to human CPSF-73 (Mandel et al. 2006), which has

also been suggested to have 50-30 exoribonuclease activity (Dominski et al. 2005) in

addition to its endonuclease activity.

In this chapter, we will focus on the sequence conservation, structure, and

function of the XRNs (Sects. 7.2–7.11). We will also discuss the other 50-30

exoribonucleases, including Rrp17 (Sect. 7.12) and RNase J1/CPSF-73 (Sect. 7.13).

7.2 Sequence Conservation of the XRNs

Yeast and most metazoans have two XRNs, with Xrn1 (175 kD) primarily in the

cytoplasm and Xrn2 (115 kD, more commonly known as Rat1 in yeast) primarily in

the nucleus. RAT1 is an essential gene in yeast, while deletion of XRN1 in yeast

leads to slow growth, sporulation defect, DNA recombination defect, and other

phenotypes. The plant Arabidopsis has three XRNs, two of which (AtXRN2 and

168 J.H. Chang et al.

AtXRN3) are in the nucleus, while the third (AtXRN4) is in the cytoplasm

(Kastenmayer and Green 2000). However, all three Arabidopsis XRNs are Xrn2

homologs, and a sequence homolog of Xrn1 may not exist in higher plants.

The amino acid sequences of the XRNs contain two highly conserved regions

(CR1 and CR2) in their N-terminal segment (Fig. 7.1). The sequence identity

among Xrn2 homologs for these two regions is 50–60%, while that between Xrn1

and Xrn2 homologs is about 40–50%. In comparison, conservation of sequences

outside of these two regions is much lower, especially between Xrn1 and Xrn2. In

fact, the larger size of Xrn1 is due to an extensive C-terminal segment that is absent

in Xrn2. The linker between CR1 and CR2 is also poorly conserved among the

XRNs, both in sequence and in length (Fig. 7.1). Several protease-sensitive sites

identified in S. cerevisiae Xrn1 map to the boundaries of these segments (Fig. 7.1)

(Page et al. 1998).

CR1 covers residues 1–354 of human Xrn1 and residues 1–407 of human Xrn2

(Fig. 7.1), as the latter has three small inserted segments. CR1 contains seven

strictly conserved acidic residues (Asp35, Asp86, Glu176, Glu178, Asp206,

Asp208, and Asp292 in human Xrn1), and it was recognized that these residues

may be homologous to those in the active site of several other Mg2+-dependent

nucleases (Solinger et al. 1999), even though CR1 shares little overall sequence

conservation with these other enzymes. Therefore, CR1 may have a crucial role in

the active site of the XRNs, which is supported by the fact that mutations of these

acidic residues abolish the exonuclease activity (Johnson 1997; Page et al. 1998;

Solinger et al. 1999). It is expected that the seven conserved acidic residues can

coordinate two Mg2+ ions for catalysis (Yang et al. 2006).

Fig. 7.1 Sequence conservation of XRNs. Schematic drawing of the domain organization of

human Xrn1, S. cerevisiae Xrn1, human Xrn2, S. cerevisiae Rat1, and S. pombe Rat1. The two

highly conserved regions (CR1 and CR2) are labeled. The 570-residue weakly conserved segment

in Xrn1 and a 120-residue segment in Xrn2/Rat1 are indicated. Small triangles in S. cerevisiaeXrn1 indicate protease-sensitive sites. The segment at the extreme C-terminus of these proteins is

not required for activity

7 50-30 Exoribonucleases 169

CR2 covers residues 426–595 of human Xrn1 and residues 509–679 of human

Xrn2 (Fig. 7.1). This segment appears to be unique to the XRNs, and has an

important role in defining the overall landscape of the active site of the XRNs

(see Sect. 7.9).

A 570-residue segment directly following CR2 shows weak sequence conserva-

tion among the Xrn1 enzymes (Fig. 7.1). For example, human and yeast Xrn1 share

26% sequence identity for this segment. In contrast, the remaining C-terminal

segments of the Xrn1 enzymes have little sequence conservation. This C-terminal

segment of yeast Xrn1 is dispensable for its exoribonuclease activity and in vivo

function, while the 570-residue segment, though weakly conserved, is required for

activity (Page et al. 1998).

The Xrn2 enzymes have a roughly 240-residue C-terminal segment following

CR2 (Fig. 7.1). Human Xrn2 and yeast Xrn2/Rat1 share 24% sequence identity for

this segment. The last 125 residues of S. pombe Rat1 can be deleted without

affecting its in vivo function at the permissive temperature (the truncation does

lead to a ts phenotype). Further deletions, removing the C-terminal 204 residues,

inactivated the protein (Shobuike et al. 2001).

Observations on the C-terminal deletion mutants of Xrn1 and Xrn2/Rat1

described above suggest that CR1 and CR2, while highly conserved among the

XRNs, are not sufficient for the activity of these enzymes. A segment following

CR2 (roughly 570 residues for Xrn1 and 120 residues for Xrn2/Rat1) is required for

activity, even though it is only weakly conserved.

The segment of the XRNs containing CR1 and CR2 is generally acidic in nature,

with a pI of 5.6 for this segment of yeast Xrn1. In contrast, the remaining C-terminal

segments of the Xrn1 enzymes are much more basic, with a pI of 9.4 for yeast Xrn1

(Page et al. 1998).

7.3 50-30 Exonuclease Activity of XRNs

The XRNs are Mg2+-dependent, processive 50-30 exoribonucleases (Stevens 1978,1980; Stevens and Poole 1995). Mn2+ can also support the catalytic activity of these

enzymes. They generally prefer single-stranded RNA substrates with a 50-endmonophosphate group. RNAs with a hydroxyl, cap, or triphosphate group at the

50-end are poor substrates for XRNs (Stevens 1978; Stevens and Poole 1995). YeastXrn1 and Rat1 also have weak exonuclease activity toward single-stranded DNA

(Page et al. 1998; Solinger et al. 1999; Stevens and Poole 1995). Yeast Xrn1 can

cleave G4 tetraplex DNA derived from guanine-rich sequences that are normally

found in telomeres (Liu and Gilbert 1994), while mouse Xrn1 can also cleave G4

tetraplex RNA (Bashkirov et al. 1997).

The presence of strong secondary structures in the RNA substrate can block or

stall the exoribonuclease activity of yeast Xrn1 and Rat1 (Poole and Stevens 1997;

Stevens and Poole 1995). A strong stem loop at the 50-end of the genome of

170 J.H. Chang et al.

Narnavirus 20 S RNA, a persistent virus in yeast, protects it from degradation by

Xrn1 (Esteban et al. 2008).

The exoribonuclease activity of yeast Xrn1 and Rat1 is inhibited by adenosine

30,50 bisphosphate (pAp) (Dichtl et al. 1997). Nearly 80% inhibition of both Xrn1

and Rat1 can be achieved with 1 mM pAp. The inhibition of Xrn1 is not affected by

the concentration of the RNA substrate, suggesting that pAp may not compete

against RNA. pCp and pUp are as potent as pAp in inhibiting Xrn1, while 50 or 30

AMP is essentially inactive. pAp is a byproduct of the sulfate assimilation pathway,

and is normally converted to 50 AMP and Pi by the enzyme 30,50 bisphosphatenucleotidase, Hal2/Met22 in yeast. Hal2 is inhibited by submillimolar

concentrations of Li+, and the resulting increase in cellular pAp concentration (up

to 3 mM) and the consequent inhibition of Xrn1 and Rat1 may be part of the

mechanism for Li+ toxicity in yeast. A similar mechanism may contribute to the

physiological effects of Li+ in other organisms, including the therapeutic effects of

Li+ for the treatment of various neurological diseases in humans.

The cellular functions of the XRNs are primarily linked to their exoribonuclease

activity. Therefore, these enzymes are involved in the turnover of mRNAs and

degradation of aberrant mRNAs (quality control) (Fig. 7.2). They are also involved

in the maturation (50 trimming) of ribosomal RNAs (rRNAs), small nucleolar RNAs

(snoRNAs), and others, as well as the degradation of hypomodified mature tRNAs

and spacer RNA byproducts from rRNA processing. The exoribonuclease activity

of Xrn2/Rat1 also contributes to transcription termination by nuclear RNA

polymerases I and II (Pol I and Pol II). The physiological functions of Xrn1 and

Cytoplasm

Nucleus

P bodyNucleolus mRNA

cap

5′

5′

5′3′

5′

5′

5′

5′

5′

5′

5.85

255rRNA Poly (A)

Rail1 hypomodifiedtRNA

cap Dcp1/2

Lsm1-7Dhh-1

Pat1

PTC

PTC

N N

N N

RISC

mRNA(siRNA)

Ago

Exosome

Rtt 103

CTD

Telomerase

TERRARNAPol.

Xrn1

Rat1/Xrn2

Fig. 7.2 Schematic drawing of the functions of XRNs

7 50-30 Exoribonucleases 171

Xrn2/Rat1 will be described in more detail in the following two sections, and the

functions of the plant XRNs are decribed in Sect. 7.7.

Some of the functional differences between Xrn1 and Xrn2/Rat1 are due to their

different cellular localizations. However, a nuclear-targeted Xrn1 can rescue the

lethal phenotype of rat1-1 (carrying a ts mutation in RAT1) yeast cells, suggestingthat Xrn1 can complement the essential function of Rat1 (Johnson 1997). Con-

versely, RAT1 expressed from a high copy-number plasmid, as well as Rat1 without

its nuclear localization sequence (NLS), can rescue the defects due to the loss of

Xrn1 (Johnson 1997).

The XRNs may have other functions that are independent of their exonuclease

activity. For example, they may mediate protein–protein interactions to recruit

other proteins or to be recruited by other proteins and/or RNA to proper locations

in the cell. Especially, yeast Rat1 is known to form a stable complex with Rai1

(Rat1 interacting protein 1), which can stimulate the exoribonuclease activity of

Rat1. Rat1 may also interact with other protein factors that are important for Pol II

termination, including Rtt103. Yeast Xrn1 may interact directly with microtubules.

The protein complexes for Xrn1 and Xrn2/Rat1 are described in a Sect. 7.6.

7.4 Functions of Xrn1

Xrn1 nuclease activity was first identified in yeast (Larimer et al. 1992; Stevens

1978). Later studies showed that the enzyme is identical to several other proteins

isolated based on other biochemical and functional properties (Kearsey and Kipling

1991), DNA strand exchange protein 1 including (Sep1) (Tishkoff et al. 1991),

DNA strand transferase 2 (Dst2) (Dykstra et al. 1991), Kar� enhancing mutant 1

(Kem1) (Kim et al. 1990), and radiation-resistant on 5 (Rar5) (Kipling et al. 1991).

Xrn1 may also be identical to the antiviral superkiller 1 (Ski1) protein (Johnson and

Kolodner 1995). This illustrates the various functions for this enzyme other than

RNA metabolism, such as DNA recombination, chromosome stability, microtubule

association, nuclear fusion, meiosis, telomere maintenance, and cellular senes-

cence. Defects in many of these processes are observed in cells lacking Xrn1

(Larimer and Stevens 1990).

Xrn1 homologs in S. pombe (also known as Exo II) (Kaslin and Heyer 1994) andhigher eukaryotes have also been cloned, including C. elegans (Newbury and

Woollard 2004), Drosophila (Pacman) (Till et al. 1998), mouse (Bashkirov et al.

1997), and humans (Sato et al. 1998; Shimoyama et al. 2003).

7.4.1 Functions of Xrn1 in RNA Degradation and Turnover

Xrn1 has important roles in mRNA degradation and turnover. This subject has been

reviewed extensively over the past few years (Conti and Izaurralde 2005; Doma and

172 J.H. Chang et al.

Parker 2007; Houseley and Tollervey 2009; Isken and Maquat 2007; Parker and

Song 2004), and will only be discussed briefly here, focusing on the functions of

Xrn1 in these processes. The basic mode of action is that RNAs with a 50-endmonophosphate are generated by decapping of mRNAs (possibly preceded by

deadenylation) or by endonucleolytic cleavage, which are then rapidly degraded

by Xrn1 (Fig. 7.3). The 30-50 exosome also plays an important role in mRNA

metabolism (see Chap. 9), although Xrn1 is the primary enzyme for mRNA

degradation and turnover in yeast. The rate of mRNA turnover is reduced in yeast

cells lacking Xrn1, leading to accumulation of non-polyadenylated mRNAs that

also partially lack the 50-end cap structure (Hsu and Stevens 1993).

Xrn1 is predominantly localized to cytoplasmic foci known as P-bodies

(processing bodies/GW bodies), which are the major location for mRNA decapping

and 50-30 degradation as well as for temporary storage of mRNAs during translation

repression (Kulkarni et al. 2010; Parker and Sheth 2007). Recent studies show that

decapping and Xrn1-mediated degradation of mRNAs can also occur on actively

translating ribosomes (Hu et al. 2009), as does deadenylation-independent

decapping initiated by nonsense-mediated decay (NMD) (Hu et al. 2010).

Endonucleolytic cleavage of mRNAs can be initiated by no-go decay (NGD) and

by the RNA-induced silencing complex (RISC) for RNA interference (RNAi)

(Fig. 7.3) (Orban and Izaurralde 2005). In addition, endonucleolytic cleavage

during maturational processing of many RNA precursors can produce byproducts

that are degraded by Xrn1. For example, Xrn1 degrades the internal transcribed

spacer ITS1 generated from pre-ribosomal RNA processing in yeast (Fig. 7.4)

(Stevens et al. 1991).

5′ cap ORF

Dcp1/2

Dcp1/2

Xrn1

Xrn1

Xrn1

Exosome

Exosome

Deadenylasecompex

Deadenylation dependent(mRNA turnover)

Deadenylation independent(NMD)

Endonuclease dependent(NGO, NMD, IRE1/PMR1, RNAi)

Poly (A)

Fig. 7.3 Schematic drawing of mRNA turnover and mRNA degradation pathways

7 50-30 Exoribonucleases 173

Recently, it has been found that Xrn1 and Rat1 can degrade hypomodified

mature tRNAs in yeast, in the rapid tRNA decay (RTD) pathway (Chernyakov

et al. 2008).

7.4.2 Functions of Xrn1 in RNA Maturation

Xrn1 plays a role in pre-ribosomal RNA processing and maturation, which may be

especially important in the absence of Rat1 activity in yeast. This will be discussed

in more detail in Sect. 7.5.1.

7.4.3 Functions of Xrn1 in DNA Recombinationand Chromosome Stability

Xrn1 was identified in a biochemical search for DNA recombination proteins (and

hence named Sep1 and Dst2) (Dykstra et al. 1991; Tishkoff et al. 1991). It has

homologous pairing and strand exchange activities in vitro. Yeast cells lacking

Xrn1 are defective for intrachromosomal recombination, sporulation, and trigger

Fig. 7.4 Schematic drawing of the pre-ribosomal RNA processing pathways. The extent of the

exonuclease trimming is indicated by the arrows

174 J.H. Chang et al.

arrest at pachytene stage in the meiotic cell cycle (Solinger et al. 1999; Tishkoff

et al. 1995). On the other hand, Xrn1 may not be involved in mitotic recombination

or mating-type switching.

Xrn1 was identified from a genetic screen for mutants that can enhance the

nuclear fusion defect of yeast cells carrying the kar1-1 mutation (hence named

Kem1) (Kim et al. 1990). Kem1 mutants also have reduced chromosome stability

and are hypersensitive to the microtubule-destabilizing drug benomyl. Defective

interactions with microtubules may be the basis of these phenotypes (see Sect. 7.6).

Yeast cells lacking Xrn1 also show cellular senescence and telomere shortening

(Liu et al. 1995), which may be related to the nuclease activity of this enzyme

toward G4 tetraplex DNA (Liu and Gilbert 1994).

Most of the defects in these nuclear processes (sporulation defect, arrest at

pachytene, chromosome instability) due to loss of Xrn1 can be rescued by targeting

Rat1 to the cytoplasm (Johnson 1997); consistent with the fact that Xrn1 is

primarily a cytoplasmic protein. This also suggests the possibility that the effects

of Xrn1 on these processes may not be direct.

7.4.4 Other Functions of Xrn1

Human Xrn1 may function as a tumor suppressor in osteogenic sarcoma, and its

expression level is reduced in these tumors (Zhang et al. 2002). Mouse Xrn1 is

highly expressed in testis, suggesting a functional role in gametogenesis (Shobuike

et al. 1997). Drosophila Xrn1/Pacman is required for male fertility (Zabolotskaya

et al. 2008). The expression level of Pacman is correlated with developmental

stages in Drosophila (Till et al. 1998), and C. elegans Xrn1 is critical for ventral

epithelial enclosure during embryogenesis (Newbury and Woollard 2004).

Xrn1 is also involved in host antiviral response. It can suppress viral RNA

recombination (Cheng et al. 2006), and down-regulate replication by HIV

(Chable-Bessia et al. 2009) and HCV (Jones et al. 2010).

7.5 Functions of Xrn2/Rat1

Like Xrn1, Xrn2 was first identified from several independent studies, due to its

different functions. It was found from a screen for ribonucleic acid trafficking

defects in yeast, and hence named Rat1 (Amberg et al. 1992), and from a screen

for protein translation defects (Hke1, homology to Kem1), which are more likely

due to defects in RNA processing or trafficking (Kenna et al. 1993). It was also

found to have functions in transcription activation (Tap1) (Aldrich et al. 1993; di

Segni et al. 1993).

In contrast to XRN1, RAT1 is an essential gene in yeast, although the exact

function (or the substrate) of this protein that is required for cell viability is

currently not known.

7 50-30 Exoribonucleases 175

Homologs of Rat1/Xrn2 in other organisms have also been cloned, including

S. pombe (also named Dhp1) (Shobuike et al. 2001; Sugano et al. 1994), mouse

(Dhm1) (Shobuike et al. 1995), and humans (Zhang et al. 1999).

7.5.1 Functions of Xrn2/Rat1 in RNA Processingand Degradation

Rat1 is required for 50-end trimming during the maturation of the 5.8 S and 25 S

rRNA, and Xrn1 can support this activity in the absence of Rat1 (El Hage et al.

2008; Fang et al. 2005; Fatica and Tollervey 2002; Geerlings et al. 2000; Henry

et al. 1994). The 5.8 S, 18 S and 25 S ribosomal RNAs are made in a single

transcript by Pol I in eukaryotes, which undergoes extensive endo and

exonucleolytic processing (Fig. 7.4). The primary transcript includes two external

transcribed spacers (50- and 30-ETS) and two internal transcribed spacers (ITS1 andITS2) (Fig. 7.4). Rat1/Xrn1 is involved in the degradation of a fragment of ITS1

that is released during pre-rRNA processing. Recent studies identified Rrp17 as an

independent 50-30 exoribonuclease that can also process the 50-ends of 5.8 S and

25 S rRNA (see Sect. 7.12) (Oeffinger et al. 2009).

Rat1 is required for 50-end processing of polycistronic and some intronic

snoRNAs in yeast, and Xrn1 can (at least partially) support this activity (Lee

et al. 2003; Petfalski et al. 1998; Qu et al. 1999; Villa et al. 1998). Rat1 and Xrn1

are involved in the degradation of some intron-containing unspliced pre-mRNAs

and intron lariats (Danin-Kreiselman et al. 2003). The entry sites for the XRNs are

produced by prior endonucleolytic cleavage or by debranching of the intron lariat.

Rat1 degrades telomeric repeat-containing RNA (TERRA) in yeast (Luke et al.

2008). Telomeres are transcribed by Pol II and polyadenylated, and cells lacking

Rat1 accumulate TERRA and have short telomeres. Therefore, Rat1 promotes

telomere elongation and is important for telomerase regulation.

7.5.2 Functions of Xrn2/Rat1 in RNA Polymerase TranscriptionTermination

Xrn2/Rat1 has a central role in the torpedo model for transcription termination by

RNA polymerases I and II. This area has been reviewed extensively over the past

few years (Buratowski 2005; Ghazal et al. 2009; Gilmour and Fan 2008; Luo and

Bentley 2004; Richard and Manley 2009; Rondon et al. 2009), and will only be

briefly discussed here.

The torpedo model suggests that the downstream RNA product, with a

50-monophosphate, produced by an endonucleolytic cleavage of the primary tran-

script serves as the entry point for a 50-30 exoribonuclease, which degrades

this downstream RNA, catches up to the elongating (or paused) polymerase, and

176 J.H. Chang et al.

causes transcription termination (Connelly and Manley 1988). The 50-30

exoribonuclease for Pol II termination was identified as Rat1 in yeast and Xrn2 in

mammalian cells (Kim et al. 2004; West et al. 2004). It was shown more recently

that Pol I transcription termination is also mediated by the torpedo model, with Rat1

being the 50-30 exoribonuclease for this function in yeast (Fig. 7.5) (El Hage et al.

2008; Kawauchi et al. 2008).

The molecular mechanism for how Xrn2/Rat1 brings about transcription termi-

nation once it catches up to the polymerase is still not clearly understood. Degrada-

tion of the downstream product is not sufficient for termination. Nuclear-targeted

Xrn1 can degrade the downstream product in yeast cells lacking Rat1, but nuclear

Xrn1 cannot cause Pol II termination (Luo et al. 2006). In addition, Rat1 alone is not

sufficient for Pol II termination in an in vitro transcription system (Dengl and

Cramer 2009). Therefore, other factors are also required for transcription termina-

tion by Rat1/Xrn2. The pre-mRNA 30-end processing factor Pcf11 may be impor-

tant for dismantling the polymerase elongation complex (Luo et al. 2006; West and

Proudfoot 2008; Zhang et al. 2005).

7.5.3 Other Functions of Xrn2/Rat1

Xrn2 is a candidate gene for spontaneous lung tumor susceptibility based

on a genome-wide association study in mice (Lu et al. 2010). In addition,

polymorphisms in human Xrn2 are associated with human lung cancer, and

over-expression of human Xrn2 can affect the differentiation of a leukemia cell

line (Park et al. 2007).

Fig. 7.5 Schematic drawing of the allosteric-torpedo (unified) model of Pol II termination.

Changes in the phosphorylation state of the CTD and in the body of Pol II are indicated (Modified

from Luo et al. 2006)

7 50-30 Exoribonucleases 177

7.6 Protein Partners of XRNs

Xrn1 is associated with the decapping machinery in yeast and may directly interact

with several of its components, including Dcp1/Dcp2, Pat1, Dhh1, and the Lsm1–7

complex (Coller and Parker 2004). This may facilitate the degradation of RNAs

once they are decapped by this machinery. The region(s) of Xrn1 that is required for

these interactions has not been identified.

Yeast Xrn1 interacts directly with tubulin and promotes microtubule assembly

(Interthal et al. 1995). Cells lacking Xrn1 show increased chromosome loss, defects

in spindle pole body separation and karyogamy, and hypersensitivity to benomyl

(Kim et al. 1990). The exonuclease activity of Xrn1 is not required for this

interaction (Solinger et al. 1999). The benomyl sensitivity of cells lacking Xrn1

can be rescued by targeting Rat1 to the cytoplasm, although cytoplasmic Rat1 does

not appear to be associated with microtubules (Johnson 1997).

In yeast, Rat1 has direct and strong association with Rai1, and the Rat1-Rai1

complex was first purified from S. cerevisiae extract (Stevens and Poole 1995).

A stable Rat1-Rai1 complex was also observed in S. pombe (Shobuike et al. 2001).Rai1 (45 kD) has orthologs in most eukaryotes, including plants, and the mamma-

lian homolog is known as Dom3Z (Xue et al. 2000). The sequences of these

orthologs are highly divergent, however, with only a few conserved residues. In

contrast to Rai1, Dom3Z does not appear to interact with Xrn2.

Rai1 is not essential for yeast cell viability, and does not have any nuclease

activity (Xue et al. 2000). However, Rai1 can moderately stimulate the

exoribonuclease activity of Rat1 (Xiang et al. 2009; Xue et al. 2000). This may

be due in part to the stabilization of Rat1 by Rai1. Rat1 is unstable and quickly loses

activity upon pre-incubation at 30 �C, whereas the Rat1-Rai1 complex is able to

retain most of its nuclease activity during this pre-incubation (Xue et al. 2000). Like

Rat1, Rai1 is also required for 5.8 S rRNA processing. However, while Rat1 is only

involved in the 50-end processing of this RNA, Rai1 is also needed for 30-endprocessing (Fang et al. 2005; Xue et al. 2000).

The Drosophila genome contains two homologs of Rai1/Dom3Z: CG9125 and

CG13190. CG13190, also known as Cutoff (Cuff), was first identified in a female-

sterile screen. cuffmutations affect germline cyst development, produce ventralized

eggs, and reduce female fecundity (Chen et al. 2007). Over-expressed Cuff is

localized in the cytoplasm and in perinuclear puncta, and Cuff does not interact

with Drosophila Xrn2.

S. cerevisiae also has a homolog of Rai1, Ydr370c, which is poorly conserved

with Rai1 at the sequence level (Xue et al. 2000). The function of this protein is

currently not known. Sequence analysis suggests that this homolog is restricted to

only a few of the fungal species, while most other eukaryotes contain only one

homolog of Rai1/Dom3Z.

Rtt103 (regulation of Ty1transposition 103) can interact with the Rat1-Rai1

complex in yeast (Dengl and Cramer 2009; Kim et al. 2004). Rtt103 was originally

found by a screen for mutants that increased Ty1 transposon’s mobility (Scholes

178 J.H. Chang et al.

et al. 2001). Rtt103 has a RNA Pol II carboxy-terminal domain (CTD)-interacting

domain (CID), and recognizes Ser2 phosphorylated CTD. Rtt103 may be involved

in nuclear pre-mRNA regulation (Kim et al. 2004), and it localizes at the 30-end of

transcribing genes together with Rat1-Rai1 in vivo (Kim et al. 2004) and in vitro

(Dengl and Cramer 2009).

A functional interaction between Rat1 and the pre-mRNA 30-end processing

factor Pcf11 has been suggested (Luo et al. 2006; West and Proudfoot 2008),

although currently there is no biochemical evidence for direct interaction between

these two proteins. Pcf11 may be responsible for the recruitment of Rat1 to the 30-end of pre-mRNAs and/or vice versa.

7.7 Functions of XRNs in Plants and Other Organisms

In Arabidopsis, AtXRN2 is involved in 50-end processing of 5.8 S and 25 S rRNAs

(Zakrzewska-Placzek et al. 2010), a function similar to that of Rat1. In addition,

both AtXRN2 and AtXRN3 can degrade miRNA loop and transgene for

suppressing endogenous post-transcriptional gene silencing (Gy et al. 2007).

The cytoplasmic AtXRN4 can degrade specific RNA transcripts but may not be a

general RNA degradation enzyme, in contrast to Xrn1. It degrades 30-end mRNA

products derived from miRNA-mediated cleavage (Souret et al. 2004). Mutation of

AtXRN4 leads to accumulation of decapped mRNAs that could be templates for

facilitating the RNAi pathway, and AtXRN4 may link mRNA degradation and

RNA silencing (Gazzani et al. 2004; Gregory et al. 2008). AtXRN4 also contributes

to the regulation of the ethylene response pathway (and hence is also known as

EIN5, ETHYLENE-INSENSITIVE5) (Olmedo et al. 2006; Potuschak et al. 2006).

In Trypanosoma brucei and other kinetoplastids, four XRN-related proteins havebeen identified, XRNA, XRNB, XRNC, and XRND (Li et al. 2006). XRND is

nuclear, XRNB and XRNC are cytoplasmic, and XRNA is present in both

compartments. XRNA and XRND are essential for growth, and XRNA is required

for degrading highly unstable, developmentally regulated mRNAs, while having

little effect on more stable, unregulated mRNAs (Li et al. 2006).

7.8 Overall Structure of Xrn2/Rat1

Crystal structure of the S. pombe Rat1-Rai1 complex is the first structural informa-

tion on the XRNs (Xiang et al. 2009). The structure of Rat1 indicates that CR1 and

CR2 form a single, large domain (Fig. 7.6a). CR1 is composed of a seven-stranded

(b1 through b7) mostly parallel b-sheet sandwiched by a-helices on both faces.

Strands b2 through b7 are arranged similar to those in the Rossmann fold, but with

strand b7 running in the opposite direction. A helix is inserted after b2 (aΒ) and b7(aD). CR2 contains several helices and long loops, which wrap around the base

7 50-30 Exoribonucleases 179

(N-terminal end) of the aD helix. Residues in the linker between CR1 and CR2 are

mostly disordered in the structure. The N- and C-termini of this segment are located

within 10 A of each other, suggesting that it is likely an inserted cassette between

the two conserved regions (Fig. 7.6a).

A striking feature of the S. pombe Rat1 structure is the long aD helix, with its

C-terminus projected 30-A away from the rest of the structure (Fig. 7.6a). This

feature has been named the “tower domain.” The N-terminal residues of helix aDare strongly conserved among XRNs, and they contribute to the formation of

the active site (see Sect. 7.9). The C-terminal residues of this helix are poorly

Fig. 7.6 Structure of the S. pombe Rat1–Rai1 complex. (a) Schematic drawing of the structure of

S. pombe Rat1–Rai1 complex (Xiang et al. 2009). The active site of Rat1 is indicated with the star,

and the arrow points to the opening of the Rai1 active site pocket. A bound divalent metal cation in

the active site of Rai1 is shown as a sphere. (b) Schematic drawing of the active site of S. pombeRat1. Side chains of residues in the active site are shown and labeled. Overall molecular surface of

(c) Rat1, (d) FEN-1 (Chapados et al. 2004), and (e) T4 RNase H (Devos et al. 2007). The active

site is indicated with the star

180 J.H. Chang et al.

conserved, and sequence analysis indicates that this helix is much shorter in Xrn1.

Two temperature-sensitive mutations in XRNs, P90L in Xrn1 (Page et al. 1998),

and Y657C in Rat1 (the rat1-1 mutation) (Luo et al. 2006), are located near the

N-terminal end of helix aD. Both mutations may destabilize this helix at the non-

permissive temperature, supporting the functional importance of the tower domain.

The structure of CR1 has many homologs, most of which are nucleases. These

include the FEN-1 family of endonucleases (Chapados et al. 2004; Hwang et al.

1998; Sakurai et al. 2005; Sayers and Artymiuk 1998), the 50 exonuclease from the

phage T5 (Ceska et al. 1996), RNase H from the phage T4 (Devos et al. 2007;

Mueser et al. 1996), the 50 nuclease domain of Taq DNA polymerase (Kim et al.

1995; Murali et al. 1998), and other PIN domain-containing nucleases (Clissold and

Ponting 2000; Glavan et al. 2006). The sequence conservation between Rat1 and

these other enzymes is very low, between 8% and 15%. The structural homology is

limited to strands b2-b7 in the central b-sheet and a few of the flanking helices. The

tower domain in Rat1 is equivalent to the helical clamp in A. fulgidus FEN-1

(Chapados et al. 2004) and the helical arch in T5 exonuclease (Ceska et al. 1996),

but the equivalent region is a long loop inM. jannaschii FEN-1 (Hwang et al. 1998)and is disordered in T4 RNase H (Devos et al. 2007; Mueser et al. 1996).

The Rat1 structure covers residues 1–874, which are sufficient for the activity of

this protein at the permissive temperature (Shobuike et al. 2001). The 120-residue

segment following CR2 forms three distinct structural features (Fig. 7.6a). The

N-terminal region (residues 752–840) of this segment adds four anti-parallel strands

(b8–b11) to the central b-sheet of CR1, producing a highly twisted 11-stranded

b-sheet. Residues 841–863 form a long loop that traverses the entire bottom face of

the central b-sheet of CR1. Finally, the C-terminal region of this segment (residues

864–874) forms an a-helix that interacts with helices aA and aH in CR1. Therefore,

despite being poorly conserved among XRNs, this segment has an important struc-

tural role, which may explain why it is required for the function of Rat1.

The strong sequence conservation for CR1 and CR2 suggests that these two

segments should have a similar structure in Xrn1 (with the exception of the tower

domain). On the other hand, most of the 570-residue segment following CR2 is

unique to Xrn1 and forms several distinct structural domains, as revealed by the

structure of Xrn1 (unpublished data).

7.9 Active Site of Xrn2/Rat1

The active site of Rat1 is located at the top of the central b-sheet of CR1, withcontributions from residues at the base of the aD helix (Fig. 7.6a). The seven

conserved acidic residues in CR1 form a cluster, and are located in the center of

the active site (Fig. 7.6b). In the structure of bacteriophage T4 RNase H, two metal

ions are associated with these acidic residues (Mueser et al. 1996), consistent with

the hypothesis that the two metal ions mediate the nuclease activity (Yang et al.

2006). Metal ions were not observed in the structure of Rat1, and there are some

7 50-30 Exoribonucleases 181

noticeable differences in the conformations of some of these acidic side chains

between Rat1 and T4 RNase H.

Three positively-charged (Lys111, Arg118, Arg119) and two polar (Gln114,

Gln115) residues at the base of the aD helix, as well as His61, His65, and Asn57 in

helix aB contribute their side chains to the active site (Fig. 7.6b). These residues

form a steep wall at one side of the active site, and may be important for interacting

with the phosphate backbone of the RNA substrate. Mutations of these residues, as

well as several of the conserved acidic residues, disrupt Rat1’s exonuclease

activity.

Residues in CR2 encircle the base of helix aD, but contribute few residues to the

Rat1 active site. The side-chain hydroxyl groups of Tyr627 and Tyr628 hydrogen-

bond with the acidic residues Glu205 and Asp237 in the active site, respectively,

and the side chain of Gln671 is located in the cluster of polar side chains from the

aB and aD helices (Fig. 7.6b).

However, CR2 introduces a dramatic difference in the overall landscape of the

active site of Rat1 as compared to other related nucleases. Due to the presence of

CR2, the Rat1 active site is a pocket (Fig. 7.6c), while the active sites of related

nucleases are more open (Figs. 7.6d,e). It has been suggested that the ssDNA

substrate threads through the helical arch in T5 nuclease (Ceska et al. 1996). In

T4 RNase H, a single-stranded DNA portion of its forked DNA substrate is also

bound in this region (Devos et al. 2007). However, such a binding mode would not

be possible in Rat1, as the substrate would clash with residues in CR2. This may be

the explanation why Rat1 is an exonuclease.

The poorly conserved C-terminal segment of Rat1, following CR2, is located

away from the active site and does not have any direct contributions to it. However,

this segment is important for recruiting Rai1, which can (indirectly) stimulate the

exoribonuclease activity of Rat1.

7.10 Structure of the Rat1-Rai1 Complex

The structure of the Rat1-Rai1 complex shows that Rai1 is bound on the opposite

face from the Rat1 active site (Fig. 7.6a), interacting primarily with the poorly

conserved C-terminal loop that traverses the bottom of CR1 (Xiang et al. 2009).

The Rat1-Rai1 interface buries approximately 800 A2 of surface area of each

protein, consistent with the stability of this complex. Ion-pair, hydrogen-bonding,

as well as van der Waals interactions contribute to the formation of this complex.

Mutations introduced in this interface can abolish the interaction as well as the

stimulation of Rat1 by Rai1 (Xiang et al. 2009).

Rai1 does not directly contribute to the active site of Rat1. Structural and

biochemical studies indicate that Rai1 enhances Rat10s exonuclease activity at

least in part by increasing the enzyme’s stability (Xue et al. 2000). This is also

supported by the observation that over-expressing Rai1 can rescue a temperature-

sensitive phenotype of Rat1 (Shobuike et al. 2001). On the other hand, real-time

182 J.H. Chang et al.

measurements of exoribonuclease activities of Rat1 and Rat1-Rai1 complex sug-

gest that the Rat1 enzyme is inherently less active (Sinturel et al. 2009). Therefore,

Rai1 may also indirectly help to organize the active site of Rat1. The structure of

Rat1 alone, and comparison with the Rat1–Rai1 complex, may reveal any changes

in the active site that is induced by Rai1 binding.

Residues at the Rat1–Rai1 interface are generally conserved among the fungal

proteins, consistent with the observations that Rat1 and Rai1 form tight complexes

in both S. cerevisiae and S. pombe (Shobuike et al. 2001; Stevens and Poole 1995).However, Rai1 residues that interact with Rat1 are not conserved in the mammalian

Rai1 homolog Dom3Z, and Dom3Z does not interact with mammalian Xrn2.

Therefore, the Rat1–Rai1 interaction appears to be unique to the fungal species.

Whether mammalian Xrn2 also has a protein partner that can stimulate its activity is

currently not known.

7.11 Rai1/Dom3Z and RNA 50-End Capping

An unexpected discovery from the structure of Rai1 is that it contains a large pocket

(Figs. 7.7a,b), and the few residues that are highly conserved among Rai1 orthologs

are all located in this pocket (rather than in the interface with Rat1) (Xiang et al.

2009). Moreover, three conserved acidic side chains, Glu150, Asp201, and Glu239

(S. pombe Rai1 numbering), together with the main-chain carbonyl of Leu240 and

two water molecules octahedrally coordinate a divalent cation (Mg2+ or Mn2+)

(Fig. 7.7c), and this metal ion is located near the bottom of the pocket (Fig. 7.7b).

Therefore, the structural information strongly suggests that Rai1 and its mammalian

homolog Dom3Z may have enzymatic activity of its own.

Biochemical studies demonstrate that Rai1 possesses RNA 50-end pyrophospho-hydrolase activity, being able to remove a pyrophosphate group from RNA with

50-end triphosphate (pppRNA) (Xiang et al. 2009). Such an enzyme (RppH) was

first characterized in bacteria (Deana et al. 2008), which is a member of the Nudix

family of enzymes. Rai1/Dom3Z shares neither sequence nor structural homology

with RppH. Remarkably, Rat1 can stimulate this pyrophosphohydrolase activity

of Rai1, even though the binding site is located far from the active site of Rai1

(Fig. 7.7a) (Xiang et al. 2009).

Further biochemical studies showed that Rai1 can also remove unmethylated 50-end cap of RNAs (GpppRNA) (Jiao et al. 2010). This activity is however distinct

from the classical decapping enzymes. First, Rai1 has much lower activity toward

methylated 50-end cap. Second, the product released by Rai1 is GpppN, while the

classical decapping enzymes release m7GDP. Therefore, Rai1 appears to have two

distinct enzymatic activities.

The amino acid sequence of the Drosophila Rai1 homolog Cuff contains

mutations at several of the conserved acidic residues that are important for metal

ion binding. It is possible that Cuff does not have RNA 50-end pyrophospho-

hydrolase and decapping activities.

7 50-30 Exoribonucleases 183

The biochemical activities of Rai1 suggest a physiological function for this

enzyme. Rai1 may be an mRNA 50-end capping quality checkpoint. Both Rai1

substrates (pppRNA and GpppRNA) are intermediates in the mRNA 50-end cappingpathway. mRNAs with defective 50-end capping cannot serve as template for

translation. At the same time, these defective mRNAs cannot be degraded by

Xrn1/Rat1, due to the protected 50-end. Therefore, Rai1 can remove the 50-endfrom such mRNAs, and the products can then be rapidly degraded by Xrn1/Rat1.

Studies in yeast show that mRNAs with 50-end capping defects are stabilized in

cells lacking Rai1, consistent with this 50-end capping quality checkpoint model

(Jiao et al. 2010). In addition, mRNAs with aberrant 50-end capping also accumu-

late under stress conditions (glucose deprivation or amino acid starvation) in cells

lacking Rai1. Moreover, defective capping in yeast cells is linked to enhanced

recruitment of Rat1 throughout the transcribing unit, and promotes Pol II termina-

tion upstream of the poly(A) site (Jimeno-Gonzalez et al. 2010). This suggests that

Rai1 can remove the defective cap in such conditions, which then allows Rat1 to

Fig. 7.7 Rai1 possesses an active site of its own. (a) Schematic drawing of the structure of

S. pombe Rai1 (Xiang et al. 2009). A bound divalent cation in the active site is shown as a sphere.

The arrow points to residues in the interface with Rat1. (b) Molecular surface of the active site

region of Rai1, showing a large pocket. The metal ion is located at the bottom of the pocket.

(c) Overlays of the metal ion binding site in the structure of Rai1 (in black) and mouse Dom3Z

(in gray). Residue numbers in parenthesis are for Dom3Z. The interaction between Glu192 in

Dom3Z and the metal ion is mediated by a water molecule

184 J.H. Chang et al.

function as a torpedo to induce Pol II termination before the completion of

transcription, in a mechanism equivalent to that of transcription termination at the

30-end of the pre-mRNA.

These studies provide the first demonstration of an mRNA 50-end capping

checkpoint (Jiao et al. 2010; Xiang et al. 2009). It was generally believed in the

field that 50-end capping always proceeds to completion. The data on Rai1 convinc-

ingly demonstrate such a checkpoint in yeast. Dom3Z has a conserved active site,

and it remains to be seen whether such a checkpoint also functions in metazoans.

7.12 The 50-30 Exoribonuclease Rrp17

Rrp17 (ribosomal RNA processing) is associated with pre-ribosomes and the nuclear

pore complex (Oeffinger et al. 2009). It is an independent nuclease for the 50-endtrimming of the 5.8 S and 25 S rRNAs. Rrp17 is an essential gene in yeast, and has

highly conserved orthologs in most eukaryotes. Rrp17 has 50-30 exoribonucleaseactivity, with preference for a phosphate group at the 50-end of the substrate, while

a triphosphate group or cap structure inhibits the nuclease activity. In comparison to

the XRNs, Rrp17 also has activity toward RNAs with a 50-end hydroxyl group. The

activity requires Mg2+ ions, while the enzyme is inactive with Mn2+.

7.13 RNase J1/CPSF-73

Earlier studies have only identified 50-30 exoribonucleases in eukaryotes, leading tothe general belief that these enzymes are not present in prokaryotes. However, it

was recently discovered that the B. subtilis endoribonuclease RNase J1 also has

50-30 exoribonuclease activity, establishing for the first time the presence of such

activity in bacteria (Condon 2010; Mathy et al. 2007). The exoribonuclease activity

is required for mRNA degradation and for 50-end maturation of 16 S rRNA in

B. subtilis. Structural studies show that the endo- and exonuclease activities share

the same active site, and suggest that RNase J1 may switch from an endo mode to

exo mode on the same RNA substrate (de la Sierra-Gallay et al. 2008).

The exoribonuclease activity of RNase J1 is more permissive toward 50-endmodification of the RNA substrate as compared to Xrn1 (Mathy et al. 2007). The

highest activity is observed for RNAwith a 50-endmonophosphate or 50-end hydroxylgroup, although this activity is roughly tenfold lower than that of Xrn1, leading to the

suggestion that RNase J1 may require a cofactor for full activity. RNA with a 50-endtriphosphate group can also be degraded, but with roughly fourfold weaker activity.

The activity toward RNA with a 50-end cap is even lower (Mathy et al. 2007).

RNase J1 exists in a complex with RNase J2, which is a sequence homolog of

RNase J1 but with little nuclease activity. RNase J1 homologs are found in bacteria

(but not in E. coli), archaea (Clouet-d’Orval et al. 2010; de la Sierra-Gallay et al.

7 50-30 Exoribonucleases 185

2008), and they may also be present in the chloroplasts of plants (de la Sierra-

Gallay et al. 2008).

RNase J1 is a structural homolog of CPSF-73 (Mandel et al. 2006), the endoribo-

nuclease for the cleavage step in eukaryotic pre-mRNA 30-end processing (Mandel

et al. 2008; Proudfoot 2004; Wahle and Ruegsegger 1999; Zhao et al. 1999). Recent

studies suggest that CPSF-73 may also have an exoribonuclease activity, degrading

the downstream cleavage product of histone pre-mRNAs (Dominski and Marzluff

2007; Dominski et al. 2005; Yang et al. 2009). Analogous to the RNase J1/J2

heterodimer, the CPSF complex also contains CPSF-100, an inactive sequence

homolog of CPSF-73. It may be possible that mammalian CPSF-73/CPSF-100

and B. subtilis RNase J1/J2 share a common evolutionary origin.

7.14 Perspectives

Studies over the past few years have greatly enhanced our understanding of the

structure and function of 50-30 exoribonucleases, as well as identified new proteins

that possess this activity. It is anticipated that further characterization of these

enzymes in the coming years, especially in higher eukaryotes, will lead to signifi-

cant new insights into the biological significance of these enzymes. Moreover,

studies on Rat1–Rai1 complex led to the discovery of a novel mRNA 50-endcapping quality checkpoint. There may be further exciting surprises from the

studies of these exoribonucleases and their interaction partners.

Acknowledgment This research is supported in part by grants from the NIH to LT (GM077175).

References

Aldrich TL, di Segni G, McConaughy BL, Keen NJ, Whelen S, Hall BD (1993) Structure of the

yeast TAP1 protein: dependence of transcription activation on the DNA context of the target

gene. Mol Cell Biol 13:3434–3444

Amberg DC, Goldstein AL, Cole CN (1992) Isolation and characterization of RAT1: an essential

gene of Saccharomyces cerevisiae required for the efficient nucleocytoplasmic trafficking of

mRNA. Genes Dev 6:1173–1189

Bashkirov VI, Scherthan H, Solinger JA, Buerstedde JM, Heyer W-D (1997) A mouse cytoplasmic

exoribonuclease (mXRN1p) with preference for G4 tetraplex substrates. J Cell Biol 136:761–773

Buratowski S (2005) Connections between mRNA 30 end processing and transcription termination.

Curr Opin Cell Biol 17:257–261

Ceska TA, Sayers JR, Stier G, Suck D (1996) A helical arch allowing single-stranded DNA to

thread through T5 50-exonuclease. Nature 382:90–93Chable-Bessia C, Meziane O, Latreille D, Triboulet R, Zamborlini A, Wagschal A, Jacquet J-M,

Reynes J, Levy Y, Saib A et al (2009) Suppression of HIV-1 replication by microRNA

effectors. Retrovirology 6:26

Chapados BR, Hosfield DJ, Han S, Qiu J, Yelent B, Shen B, Tainer JA (2004) Structural basis for

FEN-1 substrate specificity and PCNA-mediated activation in DNA replication and repair. Cell

116:39–50

186 J.H. Chang et al.

Chen Y, Pane A, Schupbach T (2007) Cutoff and aubergine mutations result in retrotransposon

upregulation and checkpoint activation in Drosophila. Curr Biol 17:637–642Cheng C-P, Serviene E, Nagy PD (2006) Suppression of viral RNA recombination by a host

exoribonuclease. J Virol 80:2631–2640

Chernyakov I, Whipple JM, Kotelawala L, Grayhack EJ, Phizicky EM (2008) Degradation of

several hypomodified mature tRNA species in Saccharomyces cerevisiae is mediated byMet22

and the 50-30 exonucleases Rat1 and Xrn1. Genes Dev 22:1369–1380

Clissold PM, Ponting CP (2000) PIN domains in nonsense-mediated mRNA decay and RNAi.

Curr Biol 10:R888–R890

Clouet-d’Orval B, Rinaldi D, Quentin Y, Carpousis AJ (2010) Euryarchaeal b-CASP proteins with

homology to bacterial RNase J Have 50-to 30-exoribonuclease activity. J Biol Chem

285:17574–17583

Coller J, Parker R (2004) Eukaryotic mRNA decapping. Annu Rev Biochem 73:861–890

Condon C (2010) What is the role of RNase J in mRNA turnover? RNA Biol 7:316–321

Connelly S, Manley JL (1988) A functional mRNA polyadenylation signal is required for

transcription termination by RNA polymerase II. Genes Dev 2:440–452

Conti E, Izaurralde E (2005) Nonsense-mediated mRNA decay: molecular insights and mechanis-

tic variations across species. Curr Opin Cell Biol 17:316–325

Danin-Kreiselman M, Lee CY, Chanfreau G (2003) RNase III-mediated degradation of unspliced

pre-mRNAs and lariat introns. Mol Cell 11:1279–1289

de la Sierra-Gallay IL, Zig L, Jamalli A, Putzer H (2008) Structural insights into the dual activity

of RNase J. Nat Struct Mol Biol 15:206–212

Deana A, Celesnik H, Belasco JG (2008) The bacterial enzyme RppH triggers messenger RNA

degradation by 50 pyrophosphate removal. Nature 451:355–358

Dengl S, Cramer P (2009) Torpedo nuclease Rat1 is insufficient to terminate RNA polymerase II

in vitro. J Biol Chem 284:21270–21279

Devos JM, Tomanicek SJ, Jones CE, Nossal NG, Mueser TC (2007) Crystal structure of bacterio-

phage T4 50 nuclease in complex with a branched DNA reveals how flap endonuclease-1 family

nucleases bind their substrates. J Biol Chem 282:31713–31724

di Segni G, McConaughy BL, Shapiro RA, Aldrich TL, Hall BD (1993) TAP1, a yeast gene that

activates the expression of a tRNA gene with a defective internal promoter. Mol Cell Biol

13:3424–3433

Dichtl B, Stevens A, Tollervey D (1997) Lithium toxicity in yeast is due to the inhibition of RNA

processing enzymes. EMBO J 16:7184–7195

Doma MK, Parker R (2007) RNA quality control in eukaryotes. Cell 131:660–668

Dominski Z, Marzluff WF (2007) Formation of the 30 end of histone mRNA: getting closer to the

end. Gene 396:373–390

Dominski Z, Yang X-C, Marzluff WF (2005) The polyadenylation factor CPSF-73 is involved in

histone-pre-mRNA processing. Cell 123:37–48

Dykstra CC, Kitada K, Clark AB, Hamatake RK, Sugino A (1991) Cloning and characterization of

DST2, the gene for DNA strand transfer protein beta from Saccharomyces cerevisiae. Mol Cell

Biol 11:2583–2592

El Hage A, Koper M, Kufel J, Tollervey D (2008) Efficient termination of transcription by RNA

polymerase I requires the 50 exonuclease Rat1 in yeast. Genes Dev 22:1069–1081

Esteban R, Vega L, Fujimura T (2008) 20S RNA Narnavirus defies the antiviral activity of SKI1/

XRN1 in Saccharomyces cerevisiae. J Biol Chem 283:25812–25820

Fang F, Phillips S, Butler JS (2005) Rat1p and Rai1p function with the nuclear exosome in the

processing and degradation of rRNA precursors. RNA 11:1571–1578

Fatica A, Tollervey D (2002) Making ribosomes. Curr Opin Cell Biol 14:313–318

Gazzani S, Lawrenson T, Woodward C, Headon D, Sablowski R (2004) A link between mRNA

turnover and RNA interference in Arabidopsis. Science 306:1046–1048Geerlings TH, Vos JC, Raue HA (2000) The final step in the formation of 25 S rRNA in

Saccharomyces cerevisiae is performed by 50–>30 exonucleases. RNA 6:1698–1703

7 50-30 Exoribonucleases 187

Ghazal G, Gagnon J, Jacques P-E, Landry J-R, Robert F, Elela SA (2009) Yeast RNase III triggers

polyadenylation-independent transcription termination. Mol Cell 36:99–109

Gilmour DS, Fan R (2008) Derailing the locomotive: transcription termination. J Biol Chem 283:

661–664

Glavan F, Behm-Ansmant I, Izaurralde E, Conti E (2006) Structures of the PIN domains of SMG6

and SMG5 reveal a nuclease within the mRNA surveillance complex. EMBO J 25:5117–5125

Gregory BD, O’Malley RC, Lister R, Urich MA, Tonti-Filippini J, Chen H, Millar AH, Ecker JR

(2008) A link between RNA metabolism and silencing affecting Arabidopsis development.

Dev Cell 14:854–866

Gy I, Gasciolli V, Lauressergues D, Morel J-B, Gombert J, Proux F, Proux C, Vaucheret H,

Mallory AC (2007) Arabidopsis FIERY1, XRN2, and XRN3 are endogenous RNA silencing

suppressors. Plant Cell 19:3451–3461

Henry Y, Wood H, Morrissey JP, Petfalski E, Kearsey S, Tollervey D (1994) The 50 end of yeast

5.8 S rRNA is generated by exonucleases from an upstream cleavage site. EMBO J 13:

2452–2463

Houseley J, Tollervey D (2009) The many pathways of RNA degradation. Cell 136:763–776

Hsu CL, Stevens A (1993) Yeast cells lacking 50–>30 exoribonuclease 1 contain mRNA species

that are poly(A) deficient and partially lack the 50 cap structure. Mol Cell Biol 13:4826–4835

Hu W, Sweet TJ, Chamnongpol S, Baker KE, Coller J (2009) Co-translational mRNA decay in

Saccharomyces cerevisiae. Nature 461:225–229Hu W, Petzold C, Coller J, Baker KE (2010) Nonsense-mediated mRNA decapping occurs on

polyribosomes in Saccharomyces cerevisiae. Nat Struct Mol Biol 17:244–247

Hwang KY, Baek K, Kim H-Y, Cho Y (1998) The crystal structure of flap endonuclease-1 from

Methanococcus jannaschii. Nat Struct Biol 5:707–713Interthal H, Bellocq C, Bahler J, Bashkirov VI, Edelstein S, Heyer W-D (1995) A role of Sep1

(¼Kem1, Xrn1) as a microtubule-associated protein in Saccharomyces cerevisiae. EMBO J

14:1057–1066

Isken O, Maquat LE (2007) Quality control of eukaryotic mRNA: safeguarding cells from

abnormal mRNA function. Genes Dev 21:1833–1856

Jiao X, Xiang S, Oh C-S, Martin CE, Tong L, Kiledjian M (2010) Identification of a quality-

control mechanism for mRNA 50-end capping. Nature 467:608–611

Jimeno-Gonzalez S, Haaning LL, Malagon F, Jensen TH (2010) The yeast 50-30 exonuclease rat1pfunctions during transcription elongation by RNA polymerase II. Mol Cell 37:580–587

Johnson AW (1997) Rat1p and Xrn1p are functionally interchangeable exoribonucleases that are

restricted to and required in the nucleus and cytoplasm, respectively. Mol Cell Biol 17:

6122–6130

Johnson AW, Kolodner RD (1995) Synthetic lethality of sep1 (xrn1) ski2 and sep1 (xrn1) ski3

mutants of Saccharomyces cerevisiae is independent of killer virus and suggests a general rolefor these genes in translation control. Mol Cell Biol 15:2719–2727

Jones DM, Domingues P, Targett-Adams P, McLauchlan J (2010) Comparison of U2OS and Huh-

7 cells for identifying host factors that affect hepatitis C virus RNA replication. J Gen Virol

91:2238–2248

Kaslin E, Heyer W-D (1994) A multifunctional exonuclease from vegetative Schizosac-charomyces pombe cells exhibiting in vitro strand exchange activity. J Biol Chem 269:

14094–14102

Kastenmayer JP, Green PJ (2000) Novel features of the XRN-family in Arabidopsis: evidence thatAtXRN4, one of several orthologs of nuclear Xrn2p/Rat1p, functions in the cytoplasm. Proc

Natl Acad Sci USA 97:13985–13990

Kawauchi J, Mischo H, Braglia P, Rondon A, Proudfoot NJ (2008) Budding yeast RNA

polymerases I and II employ parallel mechanisms of transcriptional termination. Genes Dev

22:1082–1092

Kearsey S, Kipling D (1991) Recombination and RNA processing: a common strand? Trends Cell

Biol 1:110–112

188 J.H. Chang et al.

Kenna M, Stevens A, McCammon M, Douglas MG (1993) An essential yeast gene with homology

to the exonuclease-encoding XRN1/KEM1 gene also encodes a protein with exoribonuclease

activity. Mol Cell Biol 13:341–350

Kim J, Ljungdahl PO, Fink GR (1990) Kem mutations affect nuclear fusion in Saccharomycescerevisiae. Genetics 126:799–812

Kim Y, Eom SH, Wang J, Lee D-S, Suh SW, Steitz TA (1995) Crystal structure of Thermusaquaticus DNA polymerase. Nature 376:612–616

Kim M, Krogan NJ, Vasiljeva L, Rando OJ, Nedea E, Greenblatt JF, Buratowski S (2004) The

yeast Rat1 exonuclease promotes transcription termination by RNA polymerase II. Nature

432:517–522

Kipling D, Tambini C, Kearsey SE (1991) Rar mutations which increase artificial chromosome

stability in Saccharomyces cerevisiae identify transcription and recombination proteins.

Nucleic Acids Res 19:1385–1391

Kulkarni M, Ozgur S, Stoecklin G (2010) On track with P-bodies. Biochem Soc Trans 38:

242–251

Larimer FW, Stevens A (1990) Disruption of the gene XRN1, coding for a 50-30 exoribonuclease,restricts yeast cell growth. Gene 95:85–90

Larimer FW, Hsu CL, Maupin MK, Stevens A (1992) Characterization of the XRN1 gene

encoding 50->30 exoribonuclease: sequence data and analysis of disparate protein and

mRNA levels of gene-disrupted yeast cells. Gene 120:51–57

Lee CY, Lee A, Chanfreau G (2003) The roles of endonucleolytic cleavage and exonucleolytic

digestion in the 50-end processing of S. cerevisiae box C/D snoRNAs. RNA 9:1362–1370

Li C-H, Irmer H, Gudjonsdottir-Planck D, Freese S, Salm H, Haile S, Estevez AM, Clayton C

(2006) Roles of a Trypanosoma brucei 50-30 exoribonuclease homolog in mRNA degradation.

RNA 12:2171–2186

Liu Z, Gilbert W (1994) The yeast KEM1 gene encodes a nuclease specific for G4 tetraplex DNA:

implication of in vivo functions for this novel DNA structure. Cell 77:1083–1092

Liu Z, Lee A, Gilbert W (1995) Gene disruption of a G4-DNA-dependent nuclease in yeast leads

to cellular senescence and telomere shortening. Proc Natl Acad Sci USA 92:6002–6006

Lu Y, Liu P, James M, Vikis HG, Liu H, Wen W, Franklin A, You M (2010) Genetic variants cis-

regulating Xrn2 expression contribute to the risk of spontaneous lung tumor. Oncogene

29:1041–1049

Luke B, Panza A, Redon S, Iglesias N, Li Z, Lingner J (2008) The Rat1p 50 to 30 exonucleasedegrades telomeric repeat-containing RNA and promotes telomere elongation in Saccharomy-ces cerevisiae. Mol Cell 32:465–477

Luo W, Bentley D (2004) A ribonucleolytic rat torpedoes RNA polymerase II. Cell 119:911–914

LuoW, Johnson AW, Bentley DL (2006) The role of Rat1 in coupling mRNA 30-end processing totranscription termination: implications for a unified allosteric-torpedo model. Genes Dev

20:954–965

Mandel CR, Kaneko S, Zhang H, Gebauer D, Vethantham V, Manley JL, Tong L (2006)

Polyadenylation factor CPSF-73 is the pre-mRNA 30-end-processing endonuclease. Nature

444:953–956

Mandel CR, Bai Y, Tong L (2008) Protein factors in pre-mRNA 30-end processing. Cell Mol Life

Sci 65:1099–1122

Mathy N, Benard L, Pellegrini O, Daou R, Wen T, Condon C (2007) 50-to-30 exoribonucleaseactivity in bacteria: role of RNase J1 in rRNA maturation and 50 stability of mRNA. Cell

129:681–692

Mueser TC, Nossal NG, Hyde CC (1996) Structure of bacteriophage T4 RNase H, a 50 to 30 RNA-DNA and DNA-DNA exonuclease with sequence similarity to the RAD2 family of eukaryotic

proteins. Cell 85:1101–1112

Murali R, Sharkey DJ, Daiss JL, Murthy HMK (1998) Crystal structure of Taq DNA polymerase in

complex with an inhibitory Fab: The Fab is directed against an intermediate in the helix-coil

dynamics of the enzyme. Proc Natl Acad Sci USA 95:12562–12567

7 50-30 Exoribonucleases 189

Newbury S, Woollard A (2004) The 50-30 exoribonuclease xrn-1 is essential for ventral epithelial

enclosure during C. elegans embryogenesis. RNA 10:59–65

Oeffinger M, Zenklusen D, Ferguson A, Wei KE, El Hage A, Tollervey D, Chait BT, Singer RH,

Rout MP (2009) Rrp17p is a eukaryotic exonuclease required for 50 end processing of pre-60S

ribosomal RNA. Mol Cell 36:768–781

Olmedo G, Guo H, Gregory BD, Nourizadeh SD, Aguilar-Henonin L, Li H, An F, Guzman P,

Ecker JR (2006) Ethylene-insensitive5 encodes a 50->30 exoribonuclease required for regula-

tion of the EIN3-targeting F-box proteins EBF1/2. Proc Natl Acad Sci USA 103:13286–13293

Orban TI, Izaurralde E (2005) Decay of mRNAs targeted by RISC requires XRN1, the Ski

complex, and the exosome. RNA 11:459–469

Page AM, Davis K, Molineux C, Kolodner RD, Johnson AW (1998) Mutational analysis of

exoribonuclease I from Saccharomyces cerevisiae. Nucleic Acids Res 26:3707–3716Park MH, Cho SA, Yoo KH, Yang MH, Ahn JY, Lee HS, Lee KE, Mun YC, Cho DH, Seong CM

et al (2007) Gene expression profile related to prognosis of acute myeloid leukemia. Oncol

Rep 18:1395–1402

Parker R, Sheth U (2007) P bodies and the control of mRNA translation and degradation. Mol Cell

25:635–646

Parker R, Song H (2004) The enzymes and control of eukaryotic mRNA turnover. Nat Struct Mol

Biol 11:121–127

Petfalski E, Dandekar T, Henry Y, Tollervey D (1998) Processing of the precursors to small

nucleolar RNAs and rRNAs requires common components. Mol Cell Biol 18:1181–1189

Poole TL, Stevens A (1997) Structural modifications of RNA influence the 50 exoribonucleolytichydrolysis by XRN1 and HKE1 of Saccharomyces cerevisiae. Biochem Biophys Res Commun

235:799–805

Potuschak T, Vansiri A, Binder BM, Lechner E, Vierstra RD, Genschik P (2006) The

exoribonuclease XRN4 is a component of the ethylene response pathway in Arabidopsis.Plant Cell 18:3047–3057

Proudfoot NJ (2004) New perspectives on connecting messenger RNA 30 end formation to

transcription. Curr Opin Cell Biol 16:272–278

Qu LH, Henras A, Lu YJ, Zhou H, Zhou WX, Zhu YQ, Zhao J, Henry L, Caizergues-Ferrer M,

Bachellerie JP (1999) Seven novel methylation guide small nucleolar RNAs are processed

from a common polycistronic transcript by Rat1p and RNase III in yeast. Mol Cell Biol 19:

1144–1158

Richard P, Manley JL (2009) Transcription termination by nuclear RNA polymerases. Genes Dev

23:1247–1269

Rondon AG, Mischo HE, Kawauchi J, Proudfoot NJ (2009) Fail-safe transcriptional termination

for protein-encoding genes in S. cerevisiae. Mol Cell 36:88–98

Sakurai S, Kitano K, Yamaguchi H, Hamada K, Okada K, Fukuda K, Uchida M, Ohtsuka E,

Morioka H, Hakoshima T (2005) Structural basis for recruitment of human flap endonuclease

1 to PCNA. EMBO J 24:683–693

Sato Y, Shimamoto A, Shobuike T, Sugimoto M, Ikeda H, Kuroda S, Furuichi Y (1998) Cloning

and characterization of human Sep1 (hSEP1) gene and cytoplasmic localization of its product.

DNA Res 5:241–246

Sayers JR, Artymiuk PJ (1998) Flexible loops and helical arches. Nat Struct Biol 5:668–670

Scholes DT, Banerjee M, Bowen B, Curcio MJ (2001) Multiple regulators of Ty1 transposition in

Saccharomyces cerevisiae have conserved roles in genomemaintenance. Genetics 159:1449–1465

Shimoyama Y, Morikawa Y, Ichihara M, Kodama Y, Fukuda N, Hayashi H, Morinaga T, Iwashita

T, Murakumo Y, Takahashi M (2003) Identification of human SEP1 as a glial cell line-derived

neurotrophic factor-inducible protein and its expression in the nervous system. Neurosci

121:899–906

Shobuike T, Sugano S, Yamashita T, Ikeda H (1995) Characterization of cDNA encoding mouse

homolog of fission yeast dhp1+ gene: structural and functional conservation. Nucleic Acids

Res 23:357–361

190 J.H. Chang et al.

Shobuike T, Sugano S, Yamashita T, Ikeda H (1997) Cloning and characterization of mouse Dhm2

cDNA, a functional homolog of budding yeast SEP1. Gene 191:161–166

Shobuike T, Tatebayashi K, Tani T, Sugano S, Ikeda H (2001) The dhp1+ gene, encoding a

putative nuclear 50->30 exoribonuclease, is required for proper chromosome segregation in

fission yeast. Nucleic Acids Res 29:1326–1333

Sinturel F, Pellegrini O, Xiang S, Tong L, Condon C, Benard L (2009) Real-time fluorescence

detection of exoribonucleases. RNA 15:2057–2062

Solinger JA, Pascolini D, Heyer W-D (1999) Active-site mutations in the Xrn1p exoribonuclease

of Saccharomyces cerevisiae reveal a specific role in meiosis. Mol Cell Biol 19:5930–5942

Souret FF, Kastenmayer JP, Green PJ (2004) AtXRN4 degrades mRNA in Arabidopsis and its

substrates include selected miRNA targets. Mol Cell 15:173–183

Stevens A (1978) An exoribonuclease from Saccharomyces cerevisiae: effect of modifications of

50 end groups on the hydrolysis of substrates to 50 mononucleotides. Biochem Biophys Res

Commun 81:656–661

Stevens A (1980) Purification and characterization of a Saccharomyces cerevisiae exoribonucleasewhich yields 50-mononucleotides by a 50->30 mode of hydrolysis. J Biol Chem 255:3080–3085

Stevens A, Poole TL (1995) 50-exonuclease-2 of Saccharomyces cerevisiae. Purification and

features of ribonuclease activity with comparison to 50-exonuclease-1. J Biol Chem

270:16063–16069

Stevens A, Hsu CL, Isham KR, Larimer FW (1991) Fragments of the internal transcribed spacer 1

of pre-rRNA accumulate in Saccharomyces cerevisiae lacking 50-30 exoribonuclease 1.

J Bacteriol 173:7024–7028

Sugano S, Shobuike T, Takeda T, Sugino A, Ikeda H (1994) Molecular analysis of the dhp1+ gene

of Schizosaccharomyces pombe: an essential gene that has homology to the DST2 and RAT1

genes of Saccharomyces cerevisiae. Mol Gen Genet 243:1–8

Till DD, Linz B, Seago JE, Elgar SJ, Marujo PE, Elias ML, Arraiano CM, McClellan JA,

McCarthy JE, Newbury SF (1998) Identification and developmental expression of a 50-30

exoribonuclease from Drosophila melanogaster. Mech Dev 79:51–55

Tishkoff DX, Johnson AW, Kolodner RD (1991) Molecular and genetic analysis of the gene

encoding the Saccharomyces cerevisiae strand exchange protein Sep1. Mol Cell Biol 11:

2593–2608

Tishkoff DX, Rockmill B, Roeder GS, Kolodner RD (1995) The sep1 mutant of Saccharomycescerevisiae arrests in pachytene and is deficient in meiotic recombination. Genetics 139:

495–509

Villa T, Ceradini F, Presutti C, Bozzoni I (1998) Processing of the intron-encoded U18 small

nucleolar RNA in the yeast Saccharomyces cerevisiae relies on both exo- and endonucleolyticactivities. Mol Cell Biol 18:3376–3383

Wahle E, Ruegsegger U (1999) 30-end processing of pre-mRNA in eukaryotes. FEMS Microbiol

Rev 23:277–295

West S, Proudfoot NJ (2008) Human Pcf11 enhances degradation of RNA polymerase II-

associated nascent RNA and transcriptional termination. Nucleic Acids Res 36:905–914

West S, Gromak N, Proudfoot NJ (2004) Human 50->30 exonuclease Xrn2 promotes transcription

termination at co-transcriptional cleavage sites. Nature 432:522–525

Xiang S, Cooper-Morgan A, Jiao X, Kiledjian M, Manley JL, Tong L (2009) Structure and

function of the 50->30 exoribonuclease Rat1 and its activating partner Rai1. Nature 458:

784–788

Xue Y, Bai X, Lee I, Kallstrom G, Ho J, Brown J, Stevens A, Johnson AW (2000) Saccharomycescerevisiae RAI1 (YGL246c) is homologous to human DOM3Z and encodes a protein that

binds the nuclear exoribonuclease Rat1p. Mol Cell Biol 20:4006–4015

Yang W, Lee JY, Nowotny M (2006) Making and breaking nucleic acids: two-Mg2+-ion catalysis

and substrate specificity. Mol Cell 22:5–13

Yang X-C, Sullivan KD, Marzluff WF, Dominski Z (2009) Studies of the 50 exonuclease and

endonuclease activities of CPSF-73 in histone pre-mRNA processing. Mol Cell Biol 29:31–42

7 50-30 Exoribonucleases 191

Zabolotskaya MV, Grima DP, Lin M-D, Chou T-B, Newbury SF (2008) The 50-30 exoribonucleasePacman is required for normal male fertility and is dynamically localized in cytoplasmic

particles in Drosophila testis cells. Biochem J 416:327–335

Zakrzewska-Placzek M, Souret FF, Sobczyk GJ, Green PJ, Kufel J (2010) Arabidopsis thalianaXRN2 is required for primary cleavage in the pre-ribosomal RNA. Nucleic Acids Res

38:4487–4502

Zhang M, Yu L, Xin Y, Hu P, Fu Q, Yu C, Zhao S (1999) Cloning and mapping of the XRN2 gene

to human chromosome 20p11.1-p11.2. Genomics 59:252–254

Zhang K, Dion N, Fuchs B, Damron T, Gitelis S, Irwin R, O’Connor M, Schwartz H, Scully SP,

Rock MG et al (2002) The human homolog of yeast SEP1 is a novel candidate tumor

suppressor gene in osteogenic sarcoma. Gene 298:121–127

Zhang Z, Fu J, Gilmour DS (2005) CTD-dependent dismantling of the RNA polymerase II

elongation complex by the pre-mRNA 30-end processing factor, Pcf11. Genes Dev 19:

1572–1580

Zhao J, Hyman L, Moore CL (1999) Formation of mRNA 30 ends in eukaryotes: mechanism,

regulation, and interrelationships with other steps in mRNA synthesis. Microbiol Mol Biol Rev

63:405–445

Zuo Y, Deutscher MP (2001) Exoribonuclease superfamilies: structural analysis and phylogenetic

distribution. Nucleic Acids Res 29:1017–1026

192 J.H. Chang et al.


Recommended